Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 682
Filtrar
1.
Front Immunol ; 15: 1369849, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779681

RESUMEN

Background: Stomolophus meleagris envenomation causes severe cutaneous symptoms known as jellyfish dermatitis. The potential molecule mechanisms and treatment efficiency of dermatitis remain elusive because of the complicated venom components. The biological activity and molecular regulation mechanism of Troxerutin (TRX) was firstly examined as a potential treatment for jellyfish dermatitis. Methods: We examined the inhibit effects of the TRX on tentacle extract (TE) obtained from S. meleagris in vivo and in vitro using the mice paw swelling models and corresponding assays for Enzyme-Linked Immunosorbent Assay (ELISA) Analysis, cell counting kit-8 assay, flow cytometry, respectively. The mechanism of TRX on HaCaT cells probed the altered activity of relevant signaling pathways by RNA sequencing and verified by RT-qPCR, Western blot to further confirm protective effects of TRX against the inflammation and oxidative damage caused by TE. Results: TE significantly induced the mice paw skin toxicity and accumulation of inflammatory cytokines and reactive oxygen species in vivo and vitro. Moreover, a robust increase in the phosphorylation of mitogen-activated protein kinase (MAPKs) and nuclear factor-kappa B (NF-κB) signaling pathways was observed. While, the acute cutaneous inflammation and oxidative stress induced by TE were significantly ameliorated by TRX treatment. Notablly, TRX suppressed the phosphorylation of MAPK and NF-κB by initiating the nuclear factor erythroid 2-related factor 2 signaling pathway, which result in decreasing inflammatory cytokine release. Conclusion: TRX inhibits the major signaling pathway responsible for inducing inflammatory and oxidative damage of jellyfish dermatitis, offering a novel therapy in clinical applications.


Asunto(s)
Dermatitis , Hidroxietilrutósido , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Escifozoos , Transducción de Señal , Animales , Estrés Oxidativo/efectos de los fármacos , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Dermatitis/tratamiento farmacológico , Dermatitis/metabolismo , Dermatitis/etiología , Humanos , Hidroxietilrutósido/análogos & derivados , Hidroxietilrutósido/farmacología , Hidroxietilrutósido/uso terapéutico , Venenos de Cnidarios/farmacología , Hemo-Oxigenasa 1/metabolismo , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Citocinas/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células HaCaT , Especies Reactivas de Oxígeno/metabolismo , Proteínas de la Membrana
2.
J Cell Biol ; 223(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38587472

RESUMEN

The wound-healing process is a paradigm of the directed migration of various pools of stem cells from their niche to the site of injury where they replenish damaged cells. Two decades have elapsed since the observation that wounding activates multipotent hair follicle stem cells to infiltrate the epidermis, but the cues that coax these cells out of their niche remain unknown. Here, we report that Caspase-1, a protein classically known as an integral component of the cytosolic inflammasome, is secreted upon wounding and has a non-canonical role in the extracellular milieu. Through its caspase activation recruitment domain (CARD), Caspase-1 is sufficient to initiate the migration of hair follicle stem cells into the epidermis. Uncovering this novel function of Caspase-1 also facilitates a deeper understanding of the mechanistic basis of the epithelial hyperplasia found to accompany numerous inflammatory skin diseases.


Asunto(s)
Caspasa 1 , Dermatitis , Folículo Piloso , Células Madre , Cicatrización de Heridas , Animales , Ratones , Caspasa 1/metabolismo , Movimiento Celular , Dermatitis/metabolismo , Dermatitis/patología , Cabello , Folículo Piloso/citología , Folículo Piloso/metabolismo , Inflamación/metabolismo
3.
JCI Insight ; 9(8)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470486

RESUMEN

IL-17C is an epithelial cell-derived proinflammatory cytokine whose transcriptional regulation remains unclear. Analysis of the IL17C promoter region identified TCF4 as putative regulator, and siRNA knockdown of TCF4 in human keratinocytes (KCs) increased IL17C. IL-17C stimulation of KCs (along with IL-17A and TNF-α stimulation) decreased TCF4 and increased NFKBIZ and ZC3H12A expression in an IL-17RA/RE-dependent manner, thus creating a feedback loop. ZC3H12A (MCPIP1/Regnase-1), a transcriptional immune-response regulator, also increased following TCF4 siRNA knockdown, and siRNA knockdown of ZC3H12A decreased NFKBIZ, IL1B, IL36G, CCL20, and CXCL1, revealing a proinflammatory role for ZC3H12A. Examination of lesional skin from the KC-Tie2 inflammatory dermatitis mouse model identified decreases in TCF4 protein concomitant with increases in IL-17C and Zc3h12a that reversed following the genetic elimination of Il17c, Il17ra, and Il17re and improvement in the skin phenotype. Conversely, interference with Tcf4 in KC-Tie2 mouse skin increased Il17c and exacerbated the inflammatory skin phenotype. Together, these findings identify a role for TCF4 in the negative regulation of IL-17C, which, alone and with TNF-α and IL-17A, feed back to decrease TCF4 in an IL-17RA/RE-dependent manner. This loop is further amplified by IL-17C-TCF4 autocrine regulation of ZC3H12A and IL-17C regulation of NFKBIZ to promote self-sustaining skin inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Interleucina-17 , Queratinocitos , Receptores de Interleucina-17 , Ribonucleasas , Transducción de Señal , Factor de Transcripción 4 , Animales , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Humanos , Interleucina-17/metabolismo , Interleucina-17/genética , Ratones , Queratinocitos/metabolismo , Ribonucleasas/metabolismo , Ribonucleasas/genética , Receptores de Interleucina-17/metabolismo , Receptores de Interleucina-17/genética , Inflamación/metabolismo , Inflamación/genética , Modelos Animales de Enfermedad , Epidermis/metabolismo , Dermatitis/metabolismo , Dermatitis/genética , Dermatitis/inmunología , Dermatitis/patología , Retroalimentación Fisiológica , Regulación de la Expresión Génica
4.
Cell Death Dis ; 15(3): 180, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38429278

RESUMEN

Gasdermin E (GSDME) has recently been identified as a critical executioner to mediate pyroptosis. While epidermal keratinocytes can initiate GSDME-mediated pyroptosis, the role of keratinocyte GSDME in psoriatic dermatitis remains poorly characterized. Through analysis of GEO datasets, we found elevated GSDME levels in psoriatic lesional skin. Additionally, GSDME levels correlated with both psoriasis severity and response to biologics treatments. Single-cell RNA sequencing (scRNA-seq) from a GEO dataset revealed GSDME upregulation in keratinocytes of psoriasis patients. In the imiquimod (IMQ)-induced psoriasis-like dermatitis mouse model, both full-length and cleaved forms of caspase-3 and GSDME were elevated in the epidermis. Abnormal proliferation and differentiation of keratinocytes and dermatitis were attenuated in Gsdme-/- mice and keratinocyte-specific Gsdme conditional knockout mice after IMQ stimulation. Exposure of keratinocytes to mixed cytokines (M5), mimicking psoriatic conditions, led to GSDME cleavage. Moreover, the interaction between GSDME-FL and p65 or c-jun was significantly increased after M5 stimulation. GSDME knockdown inhibited nuclear translocation of p65 and c-jun and decreased upregulation of psoriatic inflammatory mediators such as IL1ß, CCL20, CXCL1, CXCL8, S100A8, and S100A9 in M5-challenged keratinocytes. In conclusion, GSDME in keratinocytes contributes to the pathogenesis and progression of psoriasis, potentially in a pyroptosis-independent manner by interacting and promoting translocation of p65 and c-jun. These findings suggest that keratinocyte GSDME could serve as a potential therapeutic target for psoriasis treatment.


Asunto(s)
Dermatitis , Gasderminas , Psoriasis , Animales , Humanos , Ratones , Dermatitis/metabolismo , Dermatitis/patología , Gasderminas/metabolismo , Imiquimod/efectos adversos , Inflamación/patología , Queratinocitos/patología , Psoriasis/metabolismo , Psoriasis/patología , Factor de Transcripción ReIA/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo
5.
Int Immunopharmacol ; 130: 111805, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38457930

RESUMEN

OBJECTIVE: To elucidate the mechanism of Pentraxin 3 (PTX3) in the pathogenesis of psoriasiform dermatitis using Ptx3-knockout (Ptx3-KO) background mice. METHODS: An Imiquimod (IMQ)-induced murine psoriatic model was created using Ptx3-KO (Ptx3-/-) and wild-type (Ptx3+/+) mice. Skin lesion severity and expression of inflammatory mediators (IL-6 and TNFα) were assessed using PASI score and ELISA, respectively. Cutaneous tissues from the two mice groups were subjected to histological analyses, including HE staining, Masson staining, and Immunohistochemistry (IHC). The PTX3, iNOS, COX2, and Arg1 expressions were quantified and compared between the two groups. We used RNA-seq to clarify the underlying mechanisms of the disease. Flow cytometry was used to analyze systemic Th17 cell differentiation and macrophage polarization. RESULT: The psoriatic region exhibited a higher PTX3 expression than the normal cutaneous area. Moreover, PTX3 was upregulated in HaCaT cells post-TNFα stimulation. Upon IMQ stimulation, Ptx3-/- mice displayed a lower degree of the psoriasiform dermatitis phenotype compared to Ptx3+/+ mice. Consistent with the RNA-seq results, further experiments confirmed that compared to the wild-type group, the PTX3-KO group exhibited a generally lower IL-6, TNFα, iNOS, and COX2 expression and a contrasting trend in macrophage polarization. However, no significant difference in Th17 cell activation was observed between the two groups. CONCLUSIONS: This study revealed that PTX3 was upregulated in psoriatic skin tissues and TNFα-stimulated HaCaT cells. We also discovered that PTX3 deficiency in mice ameliorated the psoriasiform dermatitis phenotype upon IMQ stimulation. Mechanistically, PTX3 exacerbates psoriasiform dermatitis by regulating macrophage polarization rather than Th17 cell differentiation.


Asunto(s)
Proteína C-Reactiva , Dermatitis , Psoriasis , Componente Amiloide P Sérico , Animales , Ratones , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Imiquimod/farmacología , Interleucina-6/metabolismo , Macrófagos/patología , Psoriasis/metabolismo , Psoriasis/patología , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Humanos , Progresión de la Enfermedad , Ratones Noqueados , Ratones Endogámicos C57BL
7.
ACS Nano ; 18(4): 3424-3437, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38227828

RESUMEN

Solar dermatitis, a form of acute radiation burn that affects the skin, results from overexposure to ultraviolet B (UVB) radiation in strong sunlight. Cell damage caused by the accumulation of reactive oxygen species (ROS) produced by UVB radiation plays an important role in UVB-induced inflammation in the skin. Here, for efficiently scavenging excess ROS, modulating the microenvironment, and alleviating solar dermatitis, a π-conjugated network polyphthalocyanine supporting a highly surface-exposed Ru active site-based artificial antioxidase (HSE-PPcRu) is designed and fabricated with excellent ROS-scavenging, antioxidant, and anti-inflammatory capabilities. In photodamaged human keratinocyte cells, HSE-PPcRu could modulate mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B signaling pathways, prevent DNA damage, suppress apoptosis, inhibit pro-inflammatory cytokine secretion, and alleviate cell damage. In vivo animal experiments reveal the higher antioxidant and anti-inflammatory efficacies of HSE-PPcRu by reversing the activation of p38 and c-Jun N-terminal kinase, inhibiting expression of cyclooxygenase-2, interleukin-6, interleukin-8, and tumor necrosis factor-α. This work not only provides an idea for alleviating solar dermatitis via catalytically scavenging ROS and modulating the microenvironment but also offers a strategy to design an intelligent conjugated network-based artificial antioxidase with a highly surface-exposed active site.


Asunto(s)
Antioxidantes , Dermatitis , Animales , Humanos , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Piel/metabolismo , Rayos Ultravioleta , Antiinflamatorios/farmacología , Dermatitis/etiología , Dermatitis/metabolismo
8.
Immunopharmacol Immunotoxicol ; 46(1): 128-137, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38059657

RESUMEN

OBJECTIVE: Hydroxychloroquine (HCQ) is a US Food and Drug Administration (FDA)-approved treatment for systemic lupus erythematosus (SLE) through inhibition of antigen presentation and subsequent reduction in T cell activation. Psoriasis relapse after antimalarial therapy have been reported in up to 18% of patients with psoriasis. Here, we explored the role of HCQ on exacerbating dermatitis utilizing an imiquimod (IMQ)-induced psoriasis-like dermatitis mouse model. METHODS: Thirty-six C57BL/6 female mice were divided into six groups: wild-type control, IMQ-Only, pre-treat HCQ (30 mg/kg and 60 mg/kg HCQ), and co-treat HCQ with IMQ (30 mg/kg and 60 mg/kg HCQ). Besides control, all were topically treated with IMQ for 5 days. Pharmacological effects and mechanisms of HCQ were assessed by clinical severity of dermatitis, histopathology, and flow cytometry. HaCaT cells were co-treated with both HCQ and recombinant IL-17A, followed by the detection of proinflammatory cytokine expression and gene profiles through enzyme-linked immunosorbent assay and next-generation sequencing. RESULTS: In the pre-treated and co-treated HCQ groups, skin redness and scaling were significantly increased compared to the IMQ-Only group, and Th17 cell expression was also upregulated. Acanthosis and CD11b+IL23+ dendritic cell (DC) infiltration were observed in the HCQ treatment group. IL-6 overexpression was detected in both the HaCaT cells and skin from the experimental mice. Psoriasis-related genes were regulated after being co-treated with HCQ and recombinant IL-17A in HaCaT cells. CONCLUSIONS: HCQ exacerbates psoriasis-like skin inflammation by increasing the expression of IL-6, stimulating DC infiltration, and promoting Th17 expression in the microenvironment of the skin. KEY MESSAGES: This study provided possible mechanisms for inducing psoriasis during HCQ treatment through an animal model.


Asunto(s)
Dermatitis , Psoriasis , Humanos , Femenino , Animales , Ratones , Imiquimod/efectos adversos , Interleucina-17 , Hidroxicloroquina/efectos adversos , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Psoriasis/inducido químicamente , Queratinocitos , Piel , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C
9.
J Eur Acad Dermatol Venereol ; 38(1): 145-156, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37669859

RESUMEN

BACKGROUND: Serine metabolism is crucial for tumour oncogenesis and immune responses. S-adenosyl methionine (SAM), a methyl donor, is typically derived from serine-driven one-carbon metabolism. However, the involvement of serine metabolism in psoriatic skin inflammation remains unclear. OBJECTIVES: To investigate the association between serine metabolism and psoriatic skin inflammation. METHODS: Clinical samples were collected from patients with psoriasis and the expression of serine biosynthesis enzymes was evaluated. The HaCaT human keratinocyte cell line was transfected with small interfering RNA (siRNA) of key enzyme or treated with inhibitors. RNA sequencing and DNA methylation assays were performed to elucidate the mechanisms underlying serine metabolism-regulated psoriatic keratinocyte inflammation. An imiquimod (IMQ)-induced psoriasis mouse model was established to determine the effect of the SAM administration on psoriatic skin inflammation. RESULTS: The expression of serine synthesis pathway enzymes, including the first rate-limiting enzyme in serine biosynthesis, phosphoglycerate dehydrogenase (PHGDH), was downregulated in the epidermal lesions of patients with psoriasis compared with that in healthy controls. Suppressing PHGDH in keratinocytes promoted the production of proinflammatory cytokines and enrichment of psoriatic-related signalling pathways, including the tumour necrosis factor-alpha (TNF-α) signalling pathway, interleukin (IL)-17 signalling pathway and NF-κB signalling pathway. In particular, PHGDH inhibition markedly promoted the secretion of IL-6 in keratinocytes with or without IL-17A, IL-22, IL-1α, oncostatin M and TNF-α (mix) stimulation. Mechanistically, PHGDH inhibition upregulated the expression of IL-6 by inhibiting SAM-dependent DNA methylation at the promoter and increasing the binding of myocyte enhancer factor 2A. Furthermore, PHGDH inhibition increased the secretion of IL-6 by increasing the activation of NF-κB via SAM inhibition. SAM treatment effectively alleviated IMQ-induced psoriasis-like skin inflammation in mice. CONCLUSIONS: Our study revealed the crucial role of PHGDH in antagonising psoriatic skin inflammation and indicated that targeting serine metabolism may represent a novel therapeutic strategy for treating psoriasis.


Asunto(s)
Dermatitis , Psoriasis , Animales , Humanos , Ratones , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Metilación de ADN , Imiquimod/uso terapéutico , Interleucina-6/metabolismo , Queratinocitos/metabolismo , Metionina , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , FN-kappa B/farmacología , FN-kappa B/uso terapéutico , Psoriasis/patología , Piel/patología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Chin J Integr Med ; 30(3): 222-229, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37597119

RESUMEN

OBJECTIVE: To determine the role of Tripterygium wilfordii multiglycoside (TGW) in the treatment of psoriatic dermatitis from a cellular immunological perspective. METHODS: Mouse models of psoriatic dermatitis were established by imiquimod (IMQ). Twelve male BALB/c mice were assigned to IMQ or IMQ+TGW groups according to a random number table. Histopathological changes in vivo were assessed by hematoxylin and eosin staining. Ratios of immune cells and cytokines in mice, as well as PAM212 cell proliferation in vitro were assessed by flow cytometry. Pro-inflammatory cytokine expression was determined using reverse transcription quantitative polymerase chain reaction. RESULTS: TGW significantly ameliorated the severity of IMQ-induced psoriasis-like mouse skin lesions and restrained the activation of CD45+ cells, neutrophils and T lymphocytes (all P<0.01). Moreover, TGW significantly attenuated keratinocytes (KCs) proliferation and downregulated the mRNA levels of inflammatory cytokines including interleukin (IL)-17A, IL-23, tumor necrosis factor α, and chemokine (C-X-C motif) ligand 1 (P<0.01 or P<0.05). Furthermore, it reduced the number of γ δ T17 cells in skin lesion of mice and draining lymph nodes (P<0.01). CONCLUSIONS: TGW improved psoriasis-like inflammation by inhibiting KCs proliferation, as well as the associated immune cells and cytokine expression. It inhibited IL-17 secretion from γ δ T cells, which improved the immune-inflammatory microenvironment of psoriasis.


Asunto(s)
Dermatitis , Psoriasis , Enfermedades de la Piel , Masculino , Animales , Ratones , Tripterygium , Psoriasis/tratamiento farmacológico , Queratinocitos , Enfermedades de la Piel/metabolismo , Citocinas/metabolismo , Imiquimod/efectos adversos , Imiquimod/metabolismo , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Piel/metabolismo
11.
Br J Dermatol ; 190(4): 536-548, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-37979162

RESUMEN

BACKGROUND: Skin barrier dysfunction may both initiate and aggravate skin inflammation. However, the mechanisms involved in the inflammation process remain largely unknown. OBJECTIVES: We sought to determine how skin barrier dysfunction enhances skin inflammation and molecular mechanisms. METHODS: Skin barrier defect mice were established by tape stripping or topical use of acetone on wildtype mice, or filaggrin deficiency. RNA-Seq was employed to analyse the differentially expressed genes in mice with skin barrier defects. Primary human keratinocytes were transfected with formylpeptide receptor (FPR)1 or protein kinase R-like endoplasmic reticulum (ER) kinase (PERK) small interfering RNA to examine the effects of these gene targets. The expressions of inflammasome NOD-like receptor (NLR)C4, epidermal barrier genes and inflammatory mediators were evaluated. RESULTS: Mechanical (tape stripping), chemical (acetone) or genetic (filaggrin deficiency) barrier disruption in mice amplified the expression of proinflammatory genes, with transcriptomic profiling revealing overexpression of formylpeptide receptor (Fpr1) in the epidermis. Treatment with the FPR1 agonist fMLP in keratinocytes upregulated the expression of the NLRC4 inflammasome and increased interleukin-1ß secretion through modulation of ER stress via the PERK-eIF2α-C/EBP homologous protein pathway. The activation of the FPR1-NLRC4 axis was also observed in skin specimens from old healthy individuals with skin barrier defect or elderly mice. Conversely, topical administration with a FPR1 antagonist, or Nlrc4 silencing, led to the normalization of barrier dysfunction and alleviation of inflammatory skin responses in vivo. CONCLUSIONS: In summary, our findings show that the FPR1-NLRC4 inflammasome axis is activated upon skin barrier disruption and may explain exaggerated inflammatory responses that are observed in disease states characterized by epidermal dysfunction. Pharmacological inhibition of FPR1 or NLRC4 represents a potential therapeutic target.


Asunto(s)
Dermatitis , Proteínas Filagrina , Animales , Humanos , Ratones , Acetona/metabolismo , Acetona/farmacología , Dermatitis/metabolismo , Epidermis/metabolismo , Inflamasomas/metabolismo , Inflamación , Queratinocitos/metabolismo , Proteínas NLR/metabolismo
12.
J Pharm Sci ; 113(2): 471-485, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37898166

RESUMEN

Tacrolimus (TRL) is used for the treatment of atopic dermatitis (AD) due to its T-cell stimulation effect. However, its significantly poor water solubility, low penetration and cytotoxicity have reduced its topical applications. Herein, tacrolimus loaded nano transfersomes (TRL-NTs) were prepared, followed by their incorporation into chitosan gel to prepare tacrolimus loaded nano transfersomal gel (TRL-NTsG). TEM analysis of the TRL-NTs was performed to check their morphology. DSC, XRD and FTIR analysis of the TRL-NTs were executed after lyophilization. Similarly, rheology, spreadability and deformability of the TRL-NTsG were investigated. In vitro release, ex vivo permeation and in vitro interaction of TRL-NTsG with keratinocytes and fibroblasts as well as their co-cultures were investigated along with their in vitro cell viability analysis. Moreover, in vivo skin deposition, ear thickness, histopathology and IgE level were also determined. Besides, 6 months stability study was also performed. Results demonstrated the uniformly distributed negatively charged nanovesicles with a mean particle size distribution of 163 nm and zeta potential of -27 mV. DSC and XRD exhibited the thermal stability and amorphous form of the drug, respectively. The TRL-NTsG showed excellent deformability, spreadability and rheological behavior. In vitro release studies exhibited an 8-fold better release of TRL from the TRL-NTsG. Similarly, 6-fold better permeation and stability of the TRL-NTsG with keratinocytes and fibroblasts as well as their co-cultures was observed. Furthermore, the ear thickness (0.6 mm) of the TRL-NTsG was found significantly reduced when compared with the untreated (1.7 mm) and TRL conventional gel treated mice (1.3 mm). The H&E staining showed no toxicity of the TRL-NTsG with significantly reduced IgE levels (120 ng/mL). The formulation was found stable for at least 6 months. These results suggested the efficacy of TRL in AD-induced animal models most importantly when incorporated in NTsG.


Asunto(s)
Dermatitis , Liposomas , Ratones , Animales , Liposomas/metabolismo , Tacrolimus , Administración Cutánea , Piel/metabolismo , Dermatitis/metabolismo , Dermatitis/patología , Inmunoglobulina E/metabolismo , Inmunoglobulina E/farmacología
13.
Int J Mol Sci ; 24(24)2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38139164

RESUMEN

Glycyrol (GC) is one natural active product. Imiquimod-induced psoriasis-like Balb/c mouse models were established. The model mice were intraperitoneally injected with cyclosporine A (CsA) and GC for 8 days followed by a series of biological detections. GC had little toxicity according to the levels of peripheral blood cells, hemoglobin, blood urea nitrogen (BUN), and serum creatinine (CRE), while CsA significantly increased the levels of BUN and CRE. GC decreased the splenic index and reduced the expressions of IL-6, IL-23, and CXCL-3 in the model mice and IL-6, CXCL-1, and CXCL-2 in the inflammatory HaCaT cells. The half inhibition concentration (IC50) of GC on HaCaT cells was 29.72 µmol/L, resulting in improved apoptosis, enhanced expressions of p21, BAX, and BIK, and reduced expressions of BCL-2. GC is an immunosuppressive agent against psoriasis-like symptoms by anti-inflammatory effects, which provides a strategy for the discovery of anti-psoriatic natural products.


Asunto(s)
Dermatitis , Psoriasis , Ratones , Animales , Interleucina-6/metabolismo , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Psoriasis/metabolismo , Dermatitis/metabolismo , Antiinflamatorios/efectos adversos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inmunosupresores/efectos adversos , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad , Piel/metabolismo , Citocinas/metabolismo
14.
Int J Mol Sci ; 24(22)2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38003532

RESUMEN

Long noncoding RNAs (lncRNAs) may contribute to the formation of psoriatic lesions. The present study's objective was to identify long lncRNA genes that are differentially expressed in patient samples of psoriasis through computational analysis techniques. By using previously published RNA sequencing data from psoriatic and healthy patients (n = 324), we analysed the differential expression of lncRNAs to determine transcripts of heightened expression. We computationally screened lncRNA transcripts as annotated by GENCODE across the human genome and compared transcription in psoriatic and healthy samples from two separate studies. We observed 54 differentially expressed genes as seen in two independent datasets collected from psoriasis and healthy patients. We also identified the differential expression of LINC01215 and LINC1206 associated with the cell cycle pathway and psoriasis pathogenesis. SH3PXD2A-AS1 was identified as a participant in the STAT3/SH3PXD2A-AS1/miR-125b/STAT3 positive feedback loop. Both the SH3PXD2A-AS1 and CERNA2 genes have already been recognised as part of the IFN-γ signalling pathway regulation. Additionally, EPHA1-AS1, CYP4Z2P and SNHG12 gene upregulation have all been previously linked to inflammatory skin diseases. Differential expression of various lncRNAs affects the pathogenesis of psoriasis. Further characterisation of lncRNAs and their functions are important for developing our understanding of psoriasis.


Asunto(s)
Dermatitis , Psoriasis , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Piel/metabolismo , Psoriasis/metabolismo , Análisis de Secuencia de ARN , Dermatitis/metabolismo , Regulación Neoplásica de la Expresión Génica
15.
Arch Microbiol ; 206(1): 3, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37991548

RESUMEN

Psoriasis is one of the common chronic inflammatory skin diseases worldwide. The skin microbiota plays a role in psoriasis through regulating skin homeostasis. However, the studies on the interactions between symbiotic microbial strains and psoriasis are limited. In this study, Staphylococcus strain XSB102 was isolated from the skin of human, which was identified as Staphylococcus warneri using VITEK2 Compact. To reveal the roles of Staphylococcus warneri on psoriasis, XSB102 were applied on the back of imiquimod-induced psoriasis-like dermatitis mice. The results indicated that it exacerbated the psoriasis and significantly increased the thickening of the epidermis. Furthermore, in vitro experiments confirmed that inactivated strain XSB102 could promote the proliferation of human epidermal keratinocytes (HaCaT) cell. However, real-time quantitative PCR and immunofluorescence results suggested that the expression of inflammatory factors such as IL-17a, IL-6, and so on were not significantly increased, while extracellular matrix related factors such as Col6a3 and TGIF2 were significantly increased after XSB102 administration. This study indicates that Staphylococcus warneri XSB102 can exacerbate psoriasis and promote keratinocyte proliferation independently of inflammatory factors, which paves the way for further exploration of the relationship between skin microbiota and psoriasis.


Asunto(s)
Dermatitis , Psoriasis , Ratones , Humanos , Animales , Imiquimod/efectos adversos , Imiquimod/metabolismo , Psoriasis/inducido químicamente , Psoriasis/metabolismo , Piel , Queratinocitos/metabolismo , Staphylococcus/genética , Proliferación Celular , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Proteínas Represoras/metabolismo , Proteínas de Homeodominio/efectos adversos , Proteínas de Homeodominio/metabolismo
16.
FASEB J ; 37(12): e23309, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37983944

RESUMEN

Ultraviolet B (UVB) radiation causes skin injury by trigging excessive calcium influx and signaling cascades in the skin keratinocytes. The heat-sensitive Ca2+ -permeable transient receptor potential vanilloid 3 (TRPV3) channels robustly expressed in the keratinocytes play an important role in skin barrier formation and wound healing. Here, we report that inhibition of cutaneous TRPV3 alleviates UVB radiation-induced skin lesions. In mouse models of ear swelling and dorsal skin injury induced by a single exposure of weak UVB radiation, TRPV3 genes and proteins were upregulated in quantitative real-time PCR and Western blot assays. In accompany with TRPV3 upregulations, the expressions of proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were also increased. Knockout of the TRPV3 gene alleviates UVB-induced ear swelling and dorsal skin inflammation. Furthermore, topical applications of two selective TRPV3 inhibitors, osthole and verbascoside, resulted in a dose-dependent attenuation of skin inflammation and lesions. Taken together, our findings demonstrate the causative role of overactive TRPV3 channel function in the development of UVB-induced skin injury. Therefore, topical inhibition of TRPV3 may hold potential therapy or prevention of UVB radiation-induced skin injury.


Asunto(s)
Dermatitis , Canales de Potencial de Receptor Transitorio , Animales , Ratones , Calor , Canales de Potencial de Receptor Transitorio/metabolismo , Canales Catiónicos TRPV/metabolismo , Ratones Noqueados , Piel/metabolismo , Queratinocitos/metabolismo , Dermatitis/metabolismo , Inflamación/metabolismo
17.
Exp Dermatol ; 32(12): 2121-2130, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37926955

RESUMEN

Psoriasis is a persistent inflammatory skin disease thought to arise as a result of the infiltration of inflammatory cells and activation of keratinocytes. Recent advances in basic research and clinical experience revealed that the interleukin (IL)-23/IL-17 axis has been identified as a major immune pathway in psoriasis. However, it remains unclear how keratinocyte factors contribute to the pathology of psoriasis. Keratinocyte proline-rich protein (KPRP) is a proline-rich insoluble protein, which is present in the epidermis and is likely to be involved in the skin barrier function. Here, to investigate the potential roles of KPRP in psoriatic skin inflammation, Kprp-modified mice were applied in the imiquimod (IMQ)-induced skin inflammation model, which develops psoriasis-like epidermal hyperplasia and cutaneous inflammation features. Then, heterozygous knockout (Kprp+/- ) but not homozygous knockout (Kprp-/- ) mice displayed attenuated skin erythema compared to control wild-type mice. In addition, RNA sequencing, quantitative PCR and/or histological analysis detected changes in the expression of several molecules related to psoriatic inflammation or keratinocyte differentiation in Kprp+/- mice, but not Kprp-/- mice. Further analysis exhibited reduced IL-17-producing γδlow T cells and amplified epidermal hyperplasia in Kprp+/- mice, which were implied to be related to decreased expression of ß-defensins and increased expression of LPAR1 (Lysophosphatidic acid receptor 1), respectively. Thus, our results imply that KPRP has the potential as a therapeutic target in psoriatic skin inflammation.


Asunto(s)
Dermatitis , Psoriasis , Ratones , Animales , Imiquimod , Interleucina-17/metabolismo , Hiperplasia/patología , Epidermis/metabolismo , Dermatitis/metabolismo , Queratinocitos/metabolismo , Psoriasis/tratamiento farmacológico , Inflamación/metabolismo , Modelos Animales de Enfermedad , Piel/metabolismo
18.
PeerJ ; 11: e15976, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37780385

RESUMEN

Rosacea is a chronic inflammatory skin disease originated from damaged skin barrier and innate/adaptive immune dysregulation. Toll-like receptors (TLRs) sense injured skin and initiate downstream inflammatory and immune responses, whose role in rosacea is not fully understood. Here, via RNA-sequencing analysis, we found that the TLR signaling pathway is the top-ranked signaling pathway enriched in rosacea skin lesions, in which TLR7 is highlighted and positively correlated with the inflammation severity of disease. In LL37-induced rosacea-like mouse models, silencing TLR7 prevented the development of rosacea-like skin inflammation. Specifically, we demonstrated that overexpressing TLR7 in keratinocytes stimulates rapamycin-sensitive mTOR complex 1 (mTORC1) pathway via NFκB signaling. Ultimately, TLR7/NFκ B/mTORC1 axis promotes the production of cytokines and chemokines, leading to the migration of CD4+T cells, which are infiltrated in the lesional skin of rosacea. Our report reveals the crucial role of TLR7 in rosacea pathogenesis and indicatesa promising candidate for rosacea treatments.


Asunto(s)
Dermatitis , Rosácea , Receptor Toll-Like 7 , Animales , Ratones , Dermatitis/metabolismo , Inflamación/metabolismo , FN-kappa B/metabolismo , Rosácea/metabolismo , Piel , Receptor Toll-Like 7/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
19.
Mol Immunol ; 163: 1-12, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37703591

RESUMEN

Psoriasis is one of the most common immune-mediated chronic inflammatory skin diseases, involving excessive proliferation of keratinocyte and infiltration of immune cells. There are many factors that cause the onset of psoriasis, so the exact pathogenesis of psoriasis still needs to be determined. High mobility group box-1 (HMGB1), a pro-inflammatory cytokine, is closely related to the pathogenesis of various inflammatory diseases. However, there are few studies investigating the effects of HMGB1 on inflammatory dermatoses. Here, we found that keratinocyte in the the IMQ-treated skin lesions of psoriasis model mice expressed more HMGB1. Notably, HMGB1 produced by keratinocyte could promote the activation of inflammatory type macrophages without affecting the polarization of anti-inflammatory type macrophages. Meanwhile, the proportion of M1 type macrophages in the skin lesions is significantly increased. Moreover, local clearance of macrophages in the skin could alleviate psoriasis like inflammation. Finally, keratinocyte-derived HMGB1 could also act on itself in turn, promoting the excessive proliferation and the mRNA expression of inflammatory cytokines of keratinocyte. Therefore, this study not only found the effect of HMGB1 on the hyperproliferation of keratinocyte, but also revealed that keratinocyte could communicate with macrophages through HMGB1, thereby facilitating macrophage inflammatory polarization. Collectively, these findings have clinical significance for the research and treatment of psoriasis, HMGB1 may become a potential target for the treatment of psoriasis.


Asunto(s)
Dermatitis , Proteína HMGB1 , Psoriasis , Animales , Ratones , Citocinas/metabolismo , Dermatitis/metabolismo , Proteína HMGB1/metabolismo , Queratinocitos , Macrófagos/metabolismo , Psoriasis/tratamiento farmacológico
20.
Int J Mol Sci ; 24(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37569318

RESUMEN

The overactivity of keratinocyte cytoplasmic signaling contributes to several cutaneous inflammatory and immune pathologies. An important emerging complement to proteins responsible for this overactivity is signal repression brought about by several proteins and protein complexes with the native role of limiting inflammation. The signaling repression by these proteins distinguishes them from transmembrane receptors, kinases, and inflammasomes, which drive inflammation. For these proteins, defects or deficiencies, whether naturally arising or in experimentally engineered skin inflammation models, have clearly linked them to maintaining keratinocytes in a non-activated state or returning cells to a post-inflamed state after a signaling event. Thus, together, these proteins help to resolve acute inflammatory responses or limit the development of chronic cutaneous inflammatory disease. We present here an integrated set of demonstrated or potentially inflammation-repressive proteins or protein complexes (linear ubiquitin chain assembly complex [LUBAC], cylindromatosis lysine 63 deubiquitinase [CYLD], tumor necrosis factor alpha-induced protein 3-interacting protein 1 [TNIP1], A20, and OTULIN) for a comprehensive view of cytoplasmic signaling highlighting protein players repressing inflammation as the needed counterpoints to signal activators and amplifiers. Ebb and flow of players on both sides of this inflammation equation would be of physiological advantage to allow acute response to damage or pathogens and yet guard against chronic inflammatory disease. Further investigation of the players responsible for repressing cytoplasmic signaling would be foundational to developing new chemical-entity pharmacologics to stabilize or enhance their function when clinical intervention is needed to restore balance.


Asunto(s)
Dermatitis , Queratinocitos , Humanos , Queratinocitos/metabolismo , Transducción de Señal/fisiología , Piel/metabolismo , Citoplasma/metabolismo , Dermatitis/metabolismo , Inflamación/metabolismo , FN-kappa B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA